Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Virol Methods ; 328: 114955, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38768869

ABSTRACT

Infectious bronchitis (IB) is an acute, highly contagious contact respiratory disease of chickens caused by infectious bronchitis virus (IBV). IBV is very prone to mutation, which brings great difficulties to the prevention and control of the disease. Therefore, there is a pressing need for a method that is fast, sensitive, specific, and convenient for detecting IBV. In this study, a real-time fluorescence-based recombinase-aided amplification (RF-RAA) method was established. Primers and probe were designed based on the conserved regions of the IBV M gene and the reaction concentrations were optimized, then the specificity, sensitivity, and reproducibility of this assay were tested. The results showed that the RF-RAA method could be completed at 39℃ within 20 min, during which the results could be interpreted visually in real-time. The RF-RAA method had good specificity, no cross-reaction with common poultry pathogens, and it detected a minimum concentration of template of 2 copies/µL for IBV. Besides, its reproducibility was stable. A total of 144 clinical samples were tested by RF-RAA and real-time quantitative PCR (qPCR), 132 samples of which were positive and 12 samples were negative, and the coincidence rate of the two methods was 100 %. In conclusion, the developed RF-RAA detection method is rapid, specific, sensitive, reproducible, and convenient, which can be utilized for laboratory detection and clinical diagnosis of IBV.


Subject(s)
Chickens , Coronavirus Infections , Infectious bronchitis virus , Nucleic Acid Amplification Techniques , Poultry Diseases , Recombinases , Sensitivity and Specificity , Infectious bronchitis virus/genetics , Infectious bronchitis virus/isolation & purification , Animals , Chickens/virology , Poultry Diseases/virology , Poultry Diseases/diagnosis , Coronavirus Infections/diagnosis , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Recombinases/metabolism , Recombinases/genetics , Reproducibility of Results , Nucleic Acid Amplification Techniques/methods , Nucleic Acid Amplification Techniques/veterinary , DNA Primers/genetics , Real-Time Polymerase Chain Reaction/methods , Real-Time Polymerase Chain Reaction/veterinary , Fluorescence , Molecular Diagnostic Techniques/methods
2.
Front Cell Dev Biol ; 10: 899869, 2022.
Article in English | MEDLINE | ID: mdl-35686052

ABSTRACT

Exosomes are membrane-enclosed vesicles secreted by cells, containing a variety of biologically active ingredients including proteins, nucleic acids and lipids. In this study, we investigated the therapeutic effects of the exosomes and underlying mechanisms in a miniature pig model of ischemia/reperfusion-induced acute kidney injury (I/R-AKI). The exosomes were extracted from cultured human umbilical cord derived mesenchymal stem cells (hUC-MSCs) and infused into a miniature pig model of I/R AKI. Our results showed that 120 min of unilateral ischemia followed by reperfusion and contralateral nephrectomy resulted in renal dysfunction, severe kidney damage, apoptosis and necroptosis. Intravenous infusion of one dose of exosomes collected from about 4 × 108 hUC-MSCs significantly improved renal function and reduced apoptosis and necroptosis. Administration of hUC-MSC exosomes also reduced the expression of some pro-inflammatory cytokines/chemokines, decreased infiltration of macrophages to the injured kidneys and suppressed the phosphorylation of nuclear factor-κB and signal transducer and activator of transcription 3, two transcriptional factors related to inflammatory regulation. Moreover, hUC-MSC exosomes could promote proliferation of renal tubular cells, angiogenesis and upregulation of Klotho and Bone Morphogenetic Protein 7, two renoprotective molecules and vascular endothelial growth factor A and its receptor. Collectively, our results suggest that injection of hUC-MSC exosomes could ameliorate I/R-AKI and accelerate renal tubular cell repair and regeneration, and that hUC-MSC exosomes may be used as a potential biological therapy for Acute kidney injury patients.

3.
Transbound Emerg Dis ; 69(5): e2832-e2851, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35717667

ABSTRACT

The Chinese IBDV novel variant (nvIBDV), belonging to the genotype A2dB1b, an emerging pathotype that can cause subclinical disease with severe, prolonged immunosuppression, poses a new threat to the poultry industry. The process of the global origin, evolution and transmission dynamics of nvIBDV, however, is poorly understood. In this study, phylogenetic trees, site substitutions of amino acid (aa) and highly accurate protein structure modelling, selection pressure, evolutionary and transmission dynamics of nvIBDV were analysed. Interestingly, nvIBDV was classified into the same genogroup with the early US antigenic variants (avIBDV) but in a new lineage with a markedly different and specific pattern of 17 aa-residual substitutions: 13 in VP2 (77D, 213N, 221K, 222T, 249K, 252I, 253Q, 254N, 284A, 286I, 299S, 318D and 323E) and four in VP1 (141I, 163V, 240E and 508K). Importantly, the aa-residues 299S and 163V may play a key role in cell binding and polymerase activity, respectively. The effective population size of the circulating avIBDV experienced two growth phases, respectively, in the years 1999-2007 (in North America) and 2015-2021 (in Asia), which is consistent with the observed trend of the epidemic outbreaks. The most recent common ancestor (tMRCA) of avIBDV most first originated in the USA and was dated around the 1970s. After its emergence, the ancestor virus of this group probably spread to China around the 1990s and the variants experienced a long-term latent circulation with the accumulation of several critical aa-residue mutations in VP2 until re-emerging in 2016. At present, central China has become the epicentre of nvIBDV spread to other parts of China and Asian countries. Importantly, a strong correlation seems to exist between the transmission patterns of virus and the flow of commercial trade of live poultry and products. These findings provide important insights into the origin, evolution and transmission of the nvIBDV and will assist in the development of programs for control strategies for these emerging viruses.


Subject(s)
Birnaviridae Infections , Infectious bursal disease virus , Poultry Diseases , Amino Acids/genetics , Animals , Birnaviridae Infections/veterinary , Chickens , Mutation , Phylogeny , Poultry Diseases/epidemiology
4.
Am J Physiol Renal Physiol ; 320(6): F1030-F1044, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33900853

ABSTRACT

Pigs represent a potentially attractive model for medical research. Similar body size and physiological patterns of kidney injury that more closely mimic those described in humans make larger animals attractive for experimentation. Using larger animals, including pigs, to investigate the pathogenesis of acute kidney injury (AKI) also serves as an experimental bridge, narrowing the gap between clinical disease and preclinical discoveries. This article compares the advantages and disadvantages of large versus small AKI animal models and provides a comprehensive overview of the development and application of porcine models of AKI induced by clinically relevant insults, including ischemia-reperfusion, sepsis, and nephrotoxin exposure. The primary focus of this review is to evaluate the use of pigs for AKI studies by current investigators, including areas where more information is needed.


Subject(s)
Acute Kidney Injury , Disease Models, Animal , Swine , Animals
5.
Front Microbiol ; 12: 628545, 2021.
Article in English | MEDLINE | ID: mdl-33584629

ABSTRACT

Clade 2.3.4.4 H5Nx highly pathogenic avian influenza viruses (HPAIVs) have caused outbreaks in poultry in the world. Some of these viruses acquired internal genes from other subtype avian influenza viruses (AIVs) such as H9 and H6 for the generation of novel reassortant viruses and continually circulated in poultry. Here, we applied a duck-origin virus DK87 and a chicken-origin virus CK66 to assess the biological characteristics of novel reassortant H5N6 HPAIVs and its pathogenesis in ducks. A genetic analysis indicated that the HA genes of the two H5N6 HPAIVs were closely related to the H5 viruses of clade 2.3.4.4 circulating in Eastern Asia and classified into H5 AIV/Eastern Asia (EA)-like lineage. Their NA genes fell into Eurasian lineage had close relationship with those of H5N6 viruses circulating in China, Laos, Vietnam, Japan, and Korea. All internal genes of DK87 were aggregated closely with H5 AIV/EA-like viruses. The internal genes (PB1, PA, NP, M, and NS) of CK66 were derived from H9N2 AIV/SH98-like viruses and the PB2 were derived from H5 AIV/EA-like viruses. These results indicate that clade 2.3.4.4 H5N6 AIVs have continually evolved and recombined with the H9N2 viruses circulating in Southern China. Pathogenicity test showed that the two viruses displayed a broader tissue distribution in ducks and caused no clinical signs. These results indicated that ducks were permissive for the replication of the chicken-origin reassortant virus CK66 without prior adaptation, but the duck-origin virus DK87-inoculated ducks showed significantly higher viral titers in some organs than the CK66-inoculated ducks at 5 day post-inoculated (DPI). The recovery of viruses from oropharyngea and cloacal swabs of contacted ducks indicated that they transmitted in native ducks by direct contact. Quantitative reverse transcription PCR (qRT-PCR) results revealed that the immune-relative genes (PRRs, IFNs, Mx-1, IL-6, and IL-8) in the lungs of inoculated ducks were expressed regardless of virus origin, but the expression of these genes was significantly higher in response to infection with the DK87 virus compared to the CK66 virus at 3 DPI. Overall, we should provide further insights into how clade 2.3.4.4 H5N6 AIVs undergo genetic and pathogenic variations to prevent outbreaks of this disease.

6.
Vet Microbiol ; 246: 108745, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32605756

ABSTRACT

The H5N6 highly pathogenic avian influenza virus (HPAIV) has been circulating in China since 2013. In this report, we describe our recent chicken experimental studies investigating the pathogenicity and transmission of four H5N6 HPAIV field strains of different origins (GS39, CK44, DK47 and CK74) and the host immune responses. Four-week-old specific-pathogen-free chickens were inoculated intranasally with one of the four H5N6 HPAIV strains (one strain per group). Among the contact chickens, the GS39 and CK74 strains caused 100 % mortality, the CK44 strain caused 80 % mortality, and the DK47 strain caused 40 % mortality. The viruses were effectively replicated in multiple tissues of the inoculated chickens, in which high viral titers were detected in virus-infected tissues, and significantly upregulated expression of immune-related genes was found in the infected chickens at 24 hpi. The chicken serum antibody levels increased from 5log2 at 7 dpe to 7.67-8log2 at 14 dpe. The major histocompatibility complex molecules were upregulated 21.22- to 32.98-fold in lungs and 5.10- to 18.47-fold in spleens. In summary, H5N6 viruses can replicate within chickens and be effectively transmitted between chickens. Our study contributes to further understanding the pathogenesis of clade 2.3.4.4 H5N6 avian influenza viruses in chickens.


Subject(s)
Antibodies, Viral/blood , Chickens/immunology , Immunity, Humoral , Influenza A virus/pathogenicity , Influenza in Birds/virology , Animals , Chickens/virology , China , Influenza A virus/classification , Influenza in Birds/immunology , Poultry Diseases/immunology , Poultry Diseases/virology , Specific Pathogen-Free Organisms , Virus Replication
7.
Front Microbiol ; 11: 1246, 2020.
Article in English | MEDLINE | ID: mdl-32595623

ABSTRACT

The protein inhibitor of the activated STAT2 (PIAS2) has been implicated in many cellular processes and can also regulate viral replication in mammals. However, the role of PIAS2 in the highly pathogenic avian influenza virus (HPAIV) H5N1 replication in ducks is still unclear. Through liquid chromatography-tandem mass spectrometry (LC-MS/MS) assay, we identified that duck PIAS2 (duPIAS2) was one protein that interacted with the nucleoprotein (NP) from the H5N1 HPAIV strain of DK212. Through confocal microscopy images and Co-IP assay, we confirmed NP could interact with duPIAS2. Overexpression of duPIAS2 in primary duck embryo fibroblast (DEF) cells was shown to promote DK212 replication, and knockdown of duPIAS2 could repress DK212 replication. We further found duPIAS2 could promote NP SUMOylation through duck SUMO1 (duSUMO1), and the potential SUMOylation sites of NP were at lysines 7, 48, and 87. Furthermore, duPIAS2 promoted the replication of DK212, here relying on the activity of its SUMO E3 ligase. Duck SENP1 (duSENP1), a deSUMOylation enzyme, could repress NP SUMOylation and also inhibit DK212 replication. Together, we identified duPIAS2 could interact with NP and that duPIAS2 promoted H5N1 HPAIV replication, which might be related to NP SUMOylation.

8.
Front Immunol ; 11: 377, 2020.
Article in English | MEDLINE | ID: mdl-32184788

ABSTRACT

In mammals, tripartite motif 32 (TRIM32) is essential for regulating host innate immune responses to viral infections. However, the antiviral effect of TRIM32 in birds has not been reported. Here, we cloned the full-length duck TRIM32 (duTRIM32) cDNA from total spleen RNA of Peking duck. DuTRIM32 consists of 682 amino acids and has 95.5% similarity in amino acid sequences with chicken TRIM32 and 84.9% similarity with human TRIM32, respectively. DuTRIM32 mRNA was found to be ubiquitously expressed in all tested tissues from healthy ducks. Overexpression of duTRIM32 significantly activated the IFN-ß promoter and upregulated the mRNA levels of IFN-ß, IRF7, and Mx, which indicates that duTRIM32 is involved in the type I IFN pathway. Furthermore, duTRIM32 was found to directly interact with duck STING (duSTING) and to contribute to the expression of IFN-ß mediated by duSTING. The mRNA level of duTRIM32 was significantly upregulated in the lungs and spleens of H5N6 highly pathogenic avian influenza virus (HPAIV) infected ducks 3 days post-infection (DPI). Furthermore, overexpression of duTRIM32 could inhibit the replication of H5N6 HPAIV in duck embryo fibroblasts (DEFs). Therefore, these results indicate that duTRIM32 is involved in the type I IFN pathway and exhibit an antiviral effect against H5N6 HPAIV infection.


Subject(s)
Avian Proteins/metabolism , Ducks , Influenza A Virus, H5N8 Subtype/physiology , Influenza in Birds/immunology , Interferon-beta/metabolism , Lung/metabolism , Transcription Factors/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Gene Expression Regulation , Virus Replication
9.
Dev Comp Immunol ; 108: 103664, 2020 07.
Article in English | MEDLINE | ID: mdl-32151676

ABSTRACT

The protein inhibitor of activated STAT (PIAS) proteins are important signal transduction modulator family and regulate the innate immune signaling pathway induced by certain transcription factors, including NF-κB, IRF3, and JAK/STAT. The PIAS protein mechanism that regulates innate immune response in mammals has been well described in the literature; however, whether the PIAS gene exists in ducks as well as the role of PIAS in duck IFN-ß expression is still unclear. Here, we cloned duck PIAS (duPIAS), finding PIAS2 could repress IFN-ß production. DuPIAS2 contains SAP-PINIT-RLD-S/T characteristic domains, and its overexpression could inhibit virus-induced IFN-ß promoter activation. Moreover, duPIAS2 interacts with duck interferon regulatory factor 7 (IRF7) and inhibits IFN-ß promoter activation induced by duck IRF7. Additionally, its inhibitory function does not rely on its SUMO E3 ligase activity but rather its C-terminal portion. The above results demonstrate that duPIAS2 is a repressor of IFN-ß production induced by duck IRF7.


Subject(s)
Ducks/immunology , Interferon Regulatory Factor-7/metabolism , Poultry Diseases/immunology , Protein Inhibitors of Activated STAT/metabolism , Animals , DEAD Box Protein 58/metabolism , Ducks/metabolism , Ducks/virology , Gene Expression Regulation/immunology , HEK293 Cells , Humans , Immunity, Innate , Influenza A virus/immunology , Interferon-beta/genetics , Interferon-beta/metabolism , Poultry Diseases/virology , Promoter Regions, Genetic , Protein Binding/immunology , Protein Domains/genetics , Protein Inhibitors of Activated STAT/genetics , Protein Inhibitors of Activated STAT/isolation & purification , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Signal Transduction/immunology , Vesiculovirus/immunology
10.
Microorganisms ; 8(2)2020 Feb 07.
Article in English | MEDLINE | ID: mdl-32046051

ABSTRACT

Since 2014, highly pathogenic avian influenza (HPAI) H5N6 viruses have circulated in waterfowls and caused human infections in China, posing significant threats to the poultry industry and the public health. However, the genetics, pathogenicity and innate immune response of H5N6 HPAIVs in geese remain largely unknown. In this study, we analyzed the genetic characteristic of the two H5N6 viruses (GS38 and DK09) isolated from apparently healthy domestic goose and duck in live poultry markets (LPMs) of Southern China in 2016. Phylogenetic analysis showed that the HA genes of the two H5N6 viruses belonged to clade 2.3.4.4 and were clustered into the MIX-like group. The MIX-like group viruses have circulated in regions such as China, Japan, Korea, and Vietnam. The NA genes of the two H5N6 viruses were classified into the Eurasian sublineage. The internal genes including PB2, PB1, PA, NP, M, and NS of the two H5N6 viruses derived from the MIX-like. Therefore, our results suggested that the two H5N6 viruses were reassortants of the H5N1 and H6N6 viruses and likely derived from the same ancestor. Additionally, we evaluated the pathogenicity and transmission of the two H5N6 viruses in domestic geese. Results showed that both the two viruses caused serious clinical symptoms in all inoculated geese and led to high mortality in these birds. Both the two viruses were transmitted efficiently to contact geese and caused lethal infection in these birds. Furthermore, we found that mRNA of pattern recognition receptors (PRRs), interferons (IFNs), and stimulated genes (ISGs) exhibited different levels of activation in the lungs and spleens of the two H5N6 viruses-inoculated geese though did not protect these birds from H5N6 HPAIVs infection. Our results suggested that the clade 2.3.4.4 waterfowl-origin H5N6 HPAIVs isolated from LPMs of Southern China could cause high mortality in geese and innate immune-related genes were involved in the geese innate immune response to H5N6 HPAIVs infection. Therefore, we should pay more attention to the evolution, pathogenic variations of these viruses and enhance virological surveillance of clade 2.3.4.4 H5N6 HPAIVs in waterfowls in China.

11.
Front Microbiol ; 10: 1782, 2019.
Article in English | MEDLINE | ID: mdl-31428075

ABSTRACT

Clade 2.3.4.4 H5 avian influenza viruses (AIVs) are widely prevalent and of significant concern to the poultry industry and public health in China. Nowadays, the clade 2.3.4.4 H5N6 virus has become a dominant AIV subtype among domestic ducks in southern China. We found that waterfowl-origin clade 2.3.4.4 H5N6 viruses (A/goose/Guangdong/16568/2016, GS16568 and A/duck/Guangdong/16873/2016, DK16873) isolated from southern China in 2016 could replicate in multiple organs of inoculated ducks. DK16873 virus caused mild infections and killed 2/5 of inoculated ducks, and GS16568 virus did not kill inoculated ducks. In addition, the two viruses could be transmitted via direct contact between ducks. DK16873 and GS16568 viruses killed 2/5 and 1/5 of contact ducks, respectively. Furthermore, ducks inoculated with the two H5N6 viruses exhibited different expressions of immune-related genes in their lungs. The expression of RIG-I, TLR3 and IL6 was significantly upregulated at 12 h post-inoculation (HPI) and most of the tested immune-related genes were significantly upregulated at 3 days post-inoculation (DPI). Notably, the expression of RIG-I and IL-6 in response to DK16873 virus was significantly higher than for GS16568 virus at 12 HPI and 3 DPI. Our research have provided helpful information about the pathogenicity, transmission and immune-related genes expression in ducks infected with new H5N6 AIVs.

12.
Transbound Emerg Dis ; 66(6): 2411-2425, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31328387

ABSTRACT

Since 2014, H5 highly pathogenic avian influenza viruses (HPAIVs) from clade 2.3.4.4 have been persistently circulating in Southern China. This has caused huge losses in the poultry industry. In this study, we analysed the genetic characteristics of seven H5N6 HPAIVs of clade 2.3.4.4 that infected birds in Southern China in 2016. Phylogenetic analysis grouped the HA, PB2, PA, M and NS genes as MIX-like, and the NA genes grouped into the Eurasian lineage. The PB1 genes of the GS24, GS25, CK46 and GS74 strains belonged to the VN 2014-like group and the others were grouped as MIX-like. The NP genes of GS24 and GS25 strains belonged to the ZJ-like group, but the others were MIX-like. Thus, these viruses came from different genotypes, and the GS24, GS25, CK46 and GS74 strains displayed genotype recombination. Additionally, our results showed that the mean death time of all chickens inoculated with 105 EID50 of CK46 or GS74 viruses was 3 and 3.38 days, respectively. The viruses replicated at high titers in all tested tissues of the inoculated chickens. They also replicated in all tested tissues of naive contact chickens, but their replication titers in some tissues were significantly different (p < 0.05). Thus, the viruses displayed high pathogenicity and variable transmission in chickens. Therefore, it is necessary to focus on the pathogenic variation and molecular evolution of H5N6 HPAIVs in order to prevent and control avian influenza in China.


Subject(s)
Influenza A virus/genetics , Influenza A virus/pathogenicity , Influenza in Birds/transmission , Animals , Chickens/virology , China , Evolution, Molecular , Genotype , Influenza A virus/classification , Phylogeny , Recombination, Genetic , Virus Replication
13.
Chin Med J (Engl) ; 132(15): 1823-1832, 2019 Aug 05.
Article in English | MEDLINE | ID: mdl-31306228

ABSTRACT

BACKGROUND: Collagen type IV (COL4)-related nephropathy includes a variety of kidney diseases that occur with or without extra-renal manifestations caused by COL4A3-5 mutations. Previous studies revealed several novel mutations, including three COL4A3 missense mutations (G619R, G801R, and C1616Y) and the COL4A3 chr:228172489delA c.4317delA p.Thr1440ProfsX87 frameshift mutation that resulted in a truncated NC1 domain (hereafter named COL4A3 c.4317delA); however, the mutation mechanisms that lead to podocyte injury remain unclear. This study aimed to further explore the mutation mechanisms that lead to podocyte injury. METHODS: Wild-type (WT) and four mutant COL4A3 segments were constructed into a lentiviral plasmid, then stably transfected into human podocytes. Real-time polymerase chain reaction and Western blotting were applied to detect endoplasmic reticulum stress (ERS)- and apoptosis-related mRNA and protein levels. Then, human podocytes were treated with MG132 (a proteasome inhibitor) and brefeldin A (a transport protein inhibitor). The human podocyte findings were verified by the establishment of a mus-Col4a3 knockout mouse monoclonal podocyte using clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9 (CRISPR/Cas9) technology. RESULTS: Our data showed that COL4A3 mRNA was significantly overexpressed in the lentivirus stably transfected podocytes. Moreover, the COL4A3 protein level was significantly increased in all groups except the COL4A3 c.4317delA group. Compared to the other test groups, the COL4A3 c.4317delA group showed excessive ERS and apoptosis. Podocytes treated with MG132 showed remarkably increased intra-cellular expression of the COL4A3 c.4317delA mutation. MG132 intervention improved higher ERS and apoptosis levels in the COL4A3 c.4317delA group. Mouse monoclonal podocytes with COL4A3 chr:82717932insA c.4852insA p.Arg1618ThrfsX4 were successfully acquired; this NC1-truncated mutation suggested a higher level of ERS and relatively remarkable level of apoptosis compared to that of the WT group. CONCLUSIONS: We demonstrated that excessive ERS and ERS-induced apoptosis were involved in the podocyte injury caused by the NC1-truncated COL4A3 mutation. Furthermore, proteasome pathway intervention might become a potential treatment for collagen type IV-related nephropathy caused by a severely truncated COL4A3 mutation.


Subject(s)
Autoantigens/genetics , Collagen Type IV/genetics , Endoplasmic Reticulum Stress/physiology , Mutation/genetics , Podocytes/metabolism , Proteasome Endopeptidase Complex/metabolism , Animals , Brefeldin A/pharmacology , Endoplasmic Reticulum Stress/genetics , Frameshift Mutation/genetics , Humans , Lentivirus/genetics , Leupeptins/pharmacology , Mice , Mice, Knockout , Mutation, Missense/genetics , Podocytes/drug effects , Proteasome Endopeptidase Complex/genetics
14.
Viruses ; 11(7)2019 07 04.
Article in English | MEDLINE | ID: mdl-31277451

ABSTRACT

Highly pathogenic avian influenza H5N6 viruses have been circulating in poultry in Asia since 2013 and producing serious diseases in chickens. Here, we analyzed the genetic properties of 10 H5N6 subtypes AIVs from geese in 2015-2016 in Guangdong province. Phylogenic analysis showed that all HA genes of the 10 viruses belonged to clade 2.3.4.4, and their genes including HA, PA, PB1, M, NP, and NS all derived from Mix-like 1 (CH, VN, LS). Their PB2 genes come from Mix-like 2 (CH, VN, JP). The NA genes were classified into a Eurasian lineage. Therefore, the 10 viruses likely originate from the same ancestor and were all recombinant viruses between different genotypes. We selected A/Goose/Guangdong/GS144/2015(H5N6) (GS144) and A/Goose/Guangdong/GS148/2016(H5N6) (GS148) viruses to inoculate 5-week-old chickens intranasally with 104 EID50/0.1 mL dose intranasally to assess their pathogenicity and transmissibility. Inoculated chickens showed that the GS144 virus caused systematic infection with a lethality of 100%, but the lethality of GS148 virus was 0%. The two viruses were efficiently transmitted to contact chickens. The lethality of GS144 and GS148 virus in contact with chickens was 87.5% and 0%, respectively, which suggests that the transmissibility of GS144 virus was stronger than GS148 virus in chickens. Thus, different H5N6 viruses from the same waterfowl can show different pathogenicity and transmissibility in chickens. Continued surveillance and characteristic analysis of the H5N6 viruses will help us to keep abreast of evolution and variation in avian influenza viruses in the future.


Subject(s)
Chickens/virology , Geese/virology , Influenza A Virus, H5N8 Subtype/classification , Influenza in Birds/transmission , Influenza in Birds/virology , Poultry Diseases/virology , Animals , Disease Models, Animal , Genotype , Influenza A Virus, H5N8 Subtype/genetics , Influenza A Virus, H5N8 Subtype/isolation & purification , Influenza A Virus, H5N8 Subtype/pathogenicity , Open Reading Frames , Phylogeny , Viral Load , Virulence
15.
Clin Exp Nephrol ; 22(6): 1315-1323, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29761241

ABSTRACT

BACKGROUND: To compare the efficacy of glucocorticoids in primary focal segmental glomerulosclerosis (pFSGS) patients with moderate proteinuria. Registered at http://www.chictr.org.cn/ , study No. ChiCTR-OPN-17012789. METHODS: pFSGS patients with urine protein between 1.0 and 3.5 g/24 h were recruited from 2006 to 2016. No decline in urine protein > 50% was observed after 2 months of run-in angiotensin-converting enzyme inhibitors/angiotensin-receptor blockers (ACEI/ARB) treatment. Patients were assigned to study group (glucocorticoids with ACEI/ARB) or control group (ACEI/ARB without glucocorticoids). Variables including 24-h urinary protein, serum albumin and serum creatinine during the trial were recorded. Remission was defined as proteinuria < 0.3 g/24 h or declined > 50%, and our composite end point as > 30% decrease of eGFR or eGFR < 30 ml/min. RESULTS: A total of 102 patients were enrolled (study group N = 52, control group N = 50), and the median follow-up time was 36 (12-117) months without significant difference between groups. During the 12-month follow-up, the remission rate was significantly higher in study group [73.1 vs 50.0% (P = 0.01)], and the initial median response time was 3 months in the study group while 6 in the control group. The end point was reached by 22.2% cases in study group, and 42.0% in control. The medium survival times were study group 72 months and control 57 (P = 0.03). Minor adverse reactions were observed in 10 patients (study group N = 8, control group N = 2). CONCLUSIONS: Additional glucocorticoids therapy is more efficacious compared to ACEI/ARB alone in the treatment of patients with pFSGS and moderate proteinuria.


Subject(s)
Angiotensin Receptor Antagonists/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Glomerulosclerosis, Focal Segmental/drug therapy , Glomerulosclerosis, Focal Segmental/physiopathology , Glucocorticoids/therapeutic use , Proteinuria/drug therapy , Adult , Angiotensin Receptor Antagonists/adverse effects , Angiotensin-Converting Enzyme Inhibitors/adverse effects , Creatinine/blood , Drug Therapy, Combination , Female , Follow-Up Studies , Glomerular Filtration Rate , Glomerulosclerosis, Focal Segmental/complications , Glucocorticoids/adverse effects , Humans , Male , Middle Aged , Prospective Studies , Proteinuria/etiology , Proteinuria/urine , Remission Induction , Serum Albumin/metabolism , Survival Rate , Time Factors
16.
Front Immunol ; 9: 789, 2018.
Article in English | MEDLINE | ID: mdl-29706970

ABSTRACT

H7N9 viruses pose a threat to human health and they are no less harmful to the poultry industry than the H5N1 avian influenza viruses. However, the pathogenesis, transmissibility, and the host immune response of the H7N9 virus in chickens and mice remain unclear. In this study, we found that H7N9 viruses replicated in multiple organs of the chicken and viral shedding persisted up to 30 days postinoculation (DPI). The viruses were efficiently transmitted between chickens through direct contact. Notably, chickens infected with H7N9 had high antibody levels throughout the entire observation period and their antibody response lasted for 30 DPI. The expression levels of the pattern-recognition receptors and pro-inflammatory cytokines were found to be significantly upregulated in the brain using quantitative real-time PCR. The expression of TLR3, TLR7, MDA5, Mx, IL-1ß, IL-6, IFN-α, and IFN-γ were also significantly different in the lungs of infected chickens. We found that the viruses isolated from these birds had low pathogenicity in mice, produced little weight loss and could only replicate in the lungs. Our findings suggested that the H7N9 viruses could replicate in chickens and mice and be efficiently transmitted between chickens, which presented a significant threat to human and poultry health.


Subject(s)
Chickens/virology , Influenza A Virus, H7N9 Subtype/pathogenicity , Influenza in Birds/transmission , Animals , Mice
17.
Emerg Infect Dis ; 23(12): 2100-2102, 2017 12.
Article in English | MEDLINE | ID: mdl-29148388
18.
Vet Microbiol ; 203: 143-148, 2017 May.
Article in English | MEDLINE | ID: mdl-28619136

ABSTRACT

H5N1, a highly pathogenic avian influenza virus (HPAIV), poses a significant threat to poultry and human health. However, currently available inactivated influenza vaccines are less efficacious against viruses that display antigenic drift. In this study, we constructed a recombinant baculovirus (BV-HMNN) expressing four conserved antigen epitopes: H5N1 hemagglutinin stem area amino acids 76-130 (HA2 76-130); three tandem repeats from the ectodomain of the conserved influenza matrix protein M2 (3M2e); nucleoprotein amino acids 55-69 (NP55-69); and nucleoprotein amino acids 380-393 (NP380-393). We evaluated the immunogenicity and protective efficacy of coimmunization with an inactivated avian influenza virus vaccine (Re6) and the recombinant baculovirus (BV-HMNN) against heterologous viral infection in specific-pathogen-free chickens. The chickens immunized with both vaccines (Re6+BV-HMNN) achieved complete protection, was significantly greater than that of chickens vaccinated with Re6 alone. BV-HMNN-supplemented vaccination also reduced viral shedding more effectively than nonsupplemented vaccination. We conclude that coimmunization with both vaccines was superior to immunization with the inactivated vaccine alone in inducing cross-protection against heterologous H5N1 virus.


Subject(s)
Baculoviridae/immunology , Chickens/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/immunology , Influenza in Birds/prevention & control , Vaccination/veterinary , Amino Acid Sequence , Animals , Antigens, Viral/immunology , Chickens/virology , Cross Protection , Epitopes/immunology , Hemagglutinins/genetics , Hemagglutinins/immunology , Influenza in Birds/virology , Poultry , Vaccines, Inactivated , Viral Matrix Proteins/genetics , Viral Matrix Proteins/immunology , Virus Shedding
19.
Vaccine ; 34(5): 622-629, 2016 Jan 27.
Article in English | MEDLINE | ID: mdl-26724200

ABSTRACT

H5N1, highly pathogenic avian influenza poses, a threat to animal and human health. Rapid changes in H5N1 viruses require periodic reformulation of the conventional strain-matched vaccines, thus emphasizing the need for a broadly protective influenza vaccine. Here, we constructed BV-Dual-3M2e-LTB, a recombinant baculovirus based on baculovirus display and BacMam technology. BV-Dual-3M2e-LTB harbors a gene cassette expressing three tandem copies of the highly conserved extracellular domain of influenza M2 protein (M2e) and the mucosal adjuvant, LTB. We showed that BV-Dual-3M2e-LTB displayed the target protein (M2e/LTB) on the baculoviral surface and expressed it in transduced mammalian cells. BV-Dual-3M2e-LTB, when delivered nasally in mice, was highly immunogenic and induced superior levels of anti-M2e IgA than the non-adjuvanted baculovirus (BV-Dual-3M2e). Importantly, after challenge with different H5N1 clades (clade 0, 2.3.2.1, 2.3.4 and 4), mice inoculated with BV-Dual-3M2e-LTB displayed improved survival and decreased lung virus shedding compared with mice inoculated with BV-Dual-3M2e. The enhanced protection from BV-Dual-3M2e-LTB is mediated by T cell immunity and is primarily based on CD8(+) T cells, while mucosal antibodies alone were insufficient for protection from lethal H5N1 challenge. These results suggest that BV-Dual-3M2e-LTB has potential to protect against a broad range of H5N1 strains thereby providing a novel direction for developing broadly protective vaccines based on cellular immunity.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cross Protection , Influenza A Virus, H5N1 Subtype , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Viral Matrix Proteins/immunology , Administration, Intranasal , Animals , Antibodies, Viral/immunology , Baculoviridae , Bronchoalveolar Lavage Fluid/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Immunity, Cellular , Immunity, Mucosal , Immunoglobulin A/immunology , Lung/virology , Mice , Mice, Inbred BALB C , Vaccines, Synthetic/immunology , Virus Shedding
SELECTION OF CITATIONS
SEARCH DETAIL
...